218 research outputs found

    Caractérisation de la mort cellulaire induite par un anticorps trifonctionnel

    Get PDF
    Le dĂ©veloppement d un cancer chez un individu immunocompĂ©tent tĂ©moigne, en partie, d un Ă©chappement tumoral au systĂšme d immunosurveillance. Par consĂ©quent, la restauration ou l induction de ces mĂ©canismes de dĂ©fense anti-tumorale est une des stratĂ©gies thĂ©rapeutiques actuelles. Un des principes de l immunothĂ©rapie est basĂ© sur l injection d anticorps ayant pour cible la cellule tumorale ou les cellules effectrices de l immunitĂ©. L efficacitĂ© anti-tumorale de ces anticorps a Ă©tĂ© considĂ©rablement amĂ©liorĂ©e par une meilleure comprĂ©hension des modes d action et des effets modulateurs de ces anticorps. Ainsi, afin d optimiser l action des effecteurs immunitaires sur les cellules tumorales, un anticorps bispĂ©cifique, trifonctionnel, le catumaxomab, capable de se lier Ă  la molĂ©cule d'adhĂ©rence des cellules Ă©pithĂ©liales (EpCAM) exprimĂ©e par les cellules tumorales et Ă  l'antigĂšne CD3 des lymphocytes T, a Ă©tĂ© dĂ©veloppĂ©, essentiellement en traitement intra-pĂ©ritonĂ©al des ascites nĂ©oplasiques rĂ©fractaires.L objectif de cette Ă©tude Ă©tait de dĂ©terminer les effets immunomodulateurs du catumaxomab sur des cellules nĂ©oplasiques exprimant EpCAM, Ă  partir de deux modĂšles expĂ©rimentaux (allogĂ©nique et autologue), de rechercher une cytotoxicitĂ© induite par la catumaxomab, et de la caractĂ©riser, notamment en analysant la prĂ©sence ou non de signaux de stress inducteurs d une mort immunogĂšne tels que l exposition membranaire de la calrĂ©ticuline par les cellules tumorales prĂ©-apoptotiques, la libĂ©ration d HMGB1 et d adĂ©nosine triphosphate (ATP) dans le milieu extra-cellulaire, responsables d une activation des lymphocytes T.En prĂ©sence de cellules EpCAM+, le catumaxomab entrainait une activation majeure des lymphocytes T (expression de CD69, CD107a, HLA-DR et PD1), stimulait une rĂ©ponse inflammatoire de type Thelper 1(Th1), et provoquait la synthĂšse d interfĂ©ron-gamma par les lymphocytes T CD8. Le catumaxomab engageait le CD16 (FcR) des cellules monocytaires et NK. De plus, sur des modĂšles allogĂ©niques, le catumaxomab, provoquait une mort cellulaire associĂ©e Ă  la libĂ©ration d ATP et induisait une mort immunogĂšne aprĂšs prĂ©-incubation dans de l oxaliplatine.Par consĂ©quent, le catumaxomab permet de moduler l environnement immunitaire dans les ascites nĂ©oplasiques, et de convertir une inflammation chronique et immunosuppressive (Th2) en une inflammation aigĂŒe et immunogĂšne (Th1). En revanche, dans ces conditions, l administration seule de catumaxomab ne semble pas dĂ©clencher de mort immunogĂšne.DiffĂ©rents moyens pourraient permettre d amĂ©liorer la cytotoxicitĂ© de cet anticorps bispĂ©cifique : (1) le combiner avec un agent anti-nĂ©oplasique tel que l oxaliplatine afin de promouvoir une mort immunogĂšne, (2) affiner son action sur le CD3 des lymphocytes en modifiant sa configuration spatiale (anticorps BiTE), (3) amplifier son affinitĂ© pour le rĂ©cepteur Fc des cellules accessoires (Fc dĂ©fucosylĂ©), (4) augmenter sa cytotoxicitĂ© en modifiant la cible dirigĂ©e contre la molĂ©cule du systĂšme immunitaire (anti-PD-1 ). Enfin, l utilisation clinique pourrait ĂȘtre facilitĂ©e en humanisant cet anticorps chimĂ©rique murin afin d Ă©viter la formation d anticorps anti-murins, dirigĂ©s contre le catumaxomab.Un essai thĂ©rapeutique de phase II dont le but est d Ă©valuer l efficacitĂ© du catumaxomab intrapĂ©ritonĂ©al aprĂšs chirurgie de cytorĂ©duction complĂšte d une carcinose gastrique, chez des patients ayant reçu en prĂ©opĂ©ratoire une chimiothĂ©rapie systĂ©mique Ă  base d oxaliplatine vient de dĂ©buter. Au cours de cette Ă©tude, nous allons valider la capacitĂ© du catumaxomab 1) Ă  induire un stress cellulaire immunogĂšne et la mort des cellules cancĂ©reuses, 2) Ă  modifier la polarisation des cellules effectrices vers une maladie inflammatoire Th1, 3) Ă  promouvoir l'expression des molĂ©cules de costimulation et TRAIL sur les cellules NK et monocytes, et corrĂ©ler ces biomarqueurs immunitaires Ă  l efficacitĂ© du traitement.The development of cancer in an immunocompetent individual reflects, in part, a tumor escape from the immunosurveillance. The tumor escape is a complex, multifactorial, in which tumor cells will evade the defense mechanisms of the host by changing their microenvironment. Therefore, restoration or induction of these defense mechanisms is one of the therapeutic strategies against cancer. One of the principles of immunotherapy is based on the injection of antibodies that target tumor cells or effector cells of immunity. The anti-tumor efficacy of these antibodies has been greatly improved by a better understanding of modes of action and modulatory effects of these antibodies.Thus, to optimize the action of immune effectors to tumor cells, a bispecific antibody, trifunctional: catumaxomab, capable of binding to the adhesion molecule of the epithelial cells (EpCAM), expressed by tumor cells and the CD3 antigen of T cells, has been developed mainly in intraperitoneal treatment of refractory malignant ascites.The objective of this study was to determine the immunomodulatory effects of catumaxomab on tumoral cells expressing EpCAM, from two experimental models (allogeneic and autologous), evaluate and characterize cytotoxicity induced by catumaxomab, and analyze the presence of stress signals inducing immunogenic cell death such as membrane exposure of calreticulin by pre-apoptotic tumor cells, release of HMGB1 and of adenosine triphosphate (ATP) in the extracellular medium, inducing a T cell activation.In the presence of EpCAM + cells, catumaxomab induced a major the activation of T cells (expression of CD69, CD107a, HLA-DR and PD1), stimulated an inflammatory response Thelper type 1 (Th1) and the synthesis of interferon-gamma by CD8 T cells. Catumaxomab committed CD16 NK cells and monocytes. More, in models allogeneic catumaxomab, caused cell death associated with ATP release and induced an immunogenic cell death after pre-incubation of oxaliplatin.Therefore, catumaxomab modulates the immune environment in malignant ascites, and convert chronic and immunosuppressive inflammation (Th2) in acute and immunogenic inflammation (Th1). However, in these conditions, catumaxomab alone does not seem to trigger immunogenic cell death.the cytotoxicity of this bispecific antibody could be enhance by different techniques: (1) combining with chemotherapy such as oxaliplatin to promote immunogenic cell death, (2) refining its action on CD3 lymphocytes by changing its spatial configuration (BiTE antibody), (3) increasing its affinity for the Fc R of accessory cells (Fc aglycosylated), (4) increasing its cytotoxicity by changing the target directed against the immune molecule (anti-PD-1 ...). Finally, the clinical use could be facilitated by this humanizing murine chimeric antibody to prevent the formation of anti-murine antibodies directed against catumaxomab.A phase II clinical trial aimed to evaluate the efficacy of intraperitoneal catumaxomab after complete cytoreductive surgery of gastric carcinomatosis in patients who received preoperative systemic chemotherapy with oxaliplatin have just started. In this study, we will validate the ability of catumaxomab 1) to induce immunogenic cell stress and death of cancer cells, 2) to change the polarization of effector cells to Th1 inflammatory disease, 3) to promote the expression of costimulatory molecules and TRAIL on NK cells and monocytes, and we will correlate these immune biomarkers to treatment efficacy.PARIS11-SCD-Bib. Ă©lectronique (914719901) / SudocSudocFranceF

    Improved retroviral suicide gene transfer in colon cancer cell lines after cell synchronization with methotrexate

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Cancer gene therapy by retroviral vectors is mainly limited by the level of transduction. Retroviral gene transfer requires target cell division. Cell synchronization, obtained by drugs inducing a reversible inhibition of DNA synthesis, could therefore be proposed to precondition target cells to retroviral gene transfer. We tested whether drug-mediated cell synchronization could enhance the transfer efficiency of a retroviral-mediated gene encoding herpes simplex virus thymidine kinase (HSV-<it>tk</it>) in two colon cancer cell lines, DHDK12 and HT29.</p> <p>Methods</p> <p>Synchronization was induced by methotrexate (MTX), aracytin (ara-C) or aphidicolin. Gene transfer efficiency was assessed by the level of HSV-TK expression. Transduced cells were driven by ganciclovir (GCV) towards apoptosis that was assessed using annexin V labeling by quantitative flow cytometry.</p> <p>Results</p> <p>DHDK12 and HT29 cells were synchronized in S phase with MTX but not ara-C or aphidicolin. In synchronized DHDK12 and HT29 cells, the HSV-TK transduction rates were 2 and 1.5-fold higher than those obtained in control cells, respectively. Furthermore, the rate of apoptosis was increased two-fold in MTX-treated DHDK12 cells after treatment with GCV.</p> <p>Conclusions</p> <p>Our findings indicate that MTX-mediated synchronization of target cells allowed a significant improvement of retroviral HSV-<it>tk </it>gene transfer, resulting in an increased cell apoptosis in response to GCV. Pharmacological control of cell cycle may thus be a useful strategy to optimize the efficiency of retroviral-mediated cancer gene therapy.</p

    Potent immunomodulatory effects of the trifunctional antibody

    No full text
    Le dĂ©veloppement d’un cancer chez un individu immunocompĂ©tent tĂ©moigne, en partie, d’un Ă©chappement tumoral au systĂšme d’immunosurveillance. Par consĂ©quent, la restauration ou l’induction de ces mĂ©canismes de dĂ©fense anti-tumorale est une des stratĂ©gies thĂ©rapeutiques actuelles. Un des principes de l’immunothĂ©rapie est basĂ© sur l’injection d’anticorps ayant pour cible la cellule tumorale ou les cellules effectrices de l’immunitĂ©. L’efficacitĂ© anti-tumorale de ces anticorps a Ă©tĂ© considĂ©rablement amĂ©liorĂ©e par une meilleure comprĂ©hension des modes d’action et des effets modulateurs de ces anticorps. Ainsi, afin d’optimiser l’action des effecteurs immunitaires sur les cellules tumorales, un anticorps bispĂ©cifique, trifonctionnel, le catumaxomab, capable de se lier Ă  la molĂ©cule d'adhĂ©rence des cellules Ă©pithĂ©liales (EpCAM) exprimĂ©e par les cellules tumorales et Ă  l'antigĂšne CD3 des lymphocytes T, a Ă©tĂ© dĂ©veloppĂ©, essentiellement en traitement intra-pĂ©ritonĂ©al des ascites nĂ©oplasiques rĂ©fractaires.L’objectif de cette Ă©tude Ă©tait de dĂ©terminer les effets immunomodulateurs du catumaxomab sur des cellules nĂ©oplasiques exprimant EpCAM, Ă  partir de deux modĂšles expĂ©rimentaux (allogĂ©nique et autologue), de rechercher une cytotoxicitĂ© induite par la catumaxomab, et de la caractĂ©riser, notamment en analysant la prĂ©sence ou non de signaux de stress inducteurs d’une mort immunogĂšne tels que l’exposition membranaire de la calrĂ©ticuline par les cellules tumorales prĂ©-apoptotiques, la libĂ©ration d’HMGB1 et d’adĂ©nosine triphosphate (ATP) dans le milieu extra-cellulaire, responsables d’une activation des lymphocytes T.En prĂ©sence de cellules EpCAM+, le catumaxomab entrainait une activation majeure des lymphocytes T (expression de CD69, CD107a, HLA-DR et PD1), stimulait une rĂ©ponse inflammatoire de type Thelper 1(Th1), et provoquait la synthĂšse d’interfĂ©ron-gamma par les lymphocytes T CD8. Le catumaxomab engageait le CD16 (FcR) des cellules monocytaires et NK. De plus, sur des modĂšles allogĂ©niques, le catumaxomab, provoquait une mort cellulaire associĂ©e Ă  la libĂ©ration d’ATP et induisait une mort immunogĂšne aprĂšs prĂ©-incubation dans de l’oxaliplatine.Par consĂ©quent, le catumaxomab permet de moduler l’environnement immunitaire dans les ascites nĂ©oplasiques, et de convertir une inflammation chronique et immunosuppressive (Th2) en une inflammation aigĂŒe et immunogĂšne (Th1). En revanche, dans ces conditions, l’administration seule de catumaxomab ne semble pas dĂ©clencher de mort immunogĂšne.DiffĂ©rents moyens pourraient permettre d’amĂ©liorer la cytotoxicitĂ© de cet anticorps bispĂ©cifique : (1) le combiner avec un agent anti-nĂ©oplasique tel que l’oxaliplatine afin de promouvoir une mort immunogĂšne, (2) affiner son action sur le CD3 des lymphocytes en modifiant sa configuration spatiale (anticorps BiTE), (3) amplifier son affinitĂ© pour le rĂ©cepteur Fcdes cellules accessoires (Fc dĂ©fucosylĂ©), (4) augmenter sa cytotoxicitĂ© en modifiant la cible dirigĂ©e contre la molĂ©cule du systĂšme immunitaire (anti-PD-1
). Enfin, l’utilisation clinique pourrait ĂȘtre facilitĂ©e en humanisant cet anticorps chimĂ©rique murin afin d’éviter la formation d’anticorps anti-murins, dirigĂ©s contre le catumaxomab.Un essai thĂ©rapeutique de phase II dont le but est d’évaluer l’efficacitĂ© du catumaxomab intrapĂ©ritonĂ©al aprĂšs chirurgie de cytorĂ©duction complĂšte d’une carcinose gastrique, chez des patients ayant reçu en prĂ©opĂ©ratoire une chimiothĂ©rapie systĂ©mique Ă  base d’oxaliplatine vient de dĂ©buter. Au cours de cette Ă©tude, nous allons valider la capacitĂ© du catumaxomab 1) Ă  induire un stress cellulaire immunogĂšne et la mort des cellules cancĂ©reuses, 2) Ă  modifier la polarisation des cellules effectrices vers une maladie inflammatoire Th1, 3) Ă  promouvoir l'expression des molĂ©cules de costimulation et TRAIL sur les cellules NK et monocytes, et corrĂ©ler ces biomarqueurs immunitaires Ă  l’efficacitĂ© du traitement.The development of cancer in an immunocompetent individual reflects, in part, a tumor escape from the immunosurveillance. The tumor escape is a complex, multifactorial, in which tumor cells will evade the defense mechanisms of the host by changing their microenvironment. Therefore, restoration or induction of these defense mechanisms is one of the therapeutic strategies against cancer. One of the principles of immunotherapy is based on the injection of antibodies that target tumor cells or effector cells of immunity. The anti-tumor efficacy of these antibodies has been greatly improved by a better understanding of modes of action and modulatory effects of these antibodies.Thus, to optimize the action of immune effectors to tumor cells, a bispecific antibody, trifunctional: catumaxomab, capable of binding to the adhesion molecule of the epithelial cells (EpCAM), expressed by tumor cells and the CD3 antigen of T cells, has been developed mainly in intraperitoneal treatment of refractory malignant ascites.The objective of this study was to determine the immunomodulatory effects of catumaxomab on tumoral cells expressing EpCAM, from two experimental models (allogeneic and autologous), evaluate and characterize cytotoxicity induced by catumaxomab, and analyze the presence of stress signals inducing immunogenic cell death such as membrane exposure of calreticulin by pre-apoptotic tumor cells, release of HMGB1 and of adenosine triphosphate (ATP) in the extracellular medium, inducing a T cell activation.In the presence of EpCAM + cells, catumaxomab induced a major the activation of T cells (expression of CD69, CD107a, HLA-DR and PD1), stimulated an inflammatory response Thelper type 1 (Th1) and the synthesis of interferon-gamma by CD8 T cells. Catumaxomab committed CD16 NK cells and monocytes. More, in models allogeneic catumaxomab, caused cell death associated with ATP release and induced an immunogenic cell death after pre-incubation of oxaliplatin.Therefore, catumaxomab modulates the immune environment in malignant ascites, and convert chronic and immunosuppressive inflammation (Th2) in acute and immunogenic inflammation (Th1). However, in these conditions, catumaxomab alone does not seem to trigger immunogenic cell death.the cytotoxicity of this bispecific antibody could be enhance by different techniques: (1) combining with chemotherapy such as oxaliplatin to promote immunogenic cell death, (2) refining its action on CD3 lymphocytes by changing its spatial configuration (BiTE antibody), (3) increasing its affinity for the FcR of accessory cells (Fc aglycosylated), (4) increasing its cytotoxicity by changing the target directed against the immune molecule (anti-PD-1 ...). Finally, the clinical use could be facilitated by this humanizing murine chimeric antibody to prevent the formation of anti-murine antibodies directed against catumaxomab.A phase II clinical trial aimed to evaluate the efficacy of intraperitoneal catumaxomab after complete cytoreductive surgery of gastric carcinomatosis in patients who received preoperative systemic chemotherapy with oxaliplatin have just started. In this study, we will validate the ability of catumaxomab 1) to induce immunogenic cell stress and death of cancer cells, 2) to change the polarization of effector cells to Th1 inflammatory disease, 3) to promote the expression of costimulatory molecules and TRAIL on NK cells and monocytes, and we will correlate these immune biomarkers to treatment efficacy

    Caractérisation de la mort cellulaire induite par un anticorps trifonctionnel

    No full text
    The development of cancer in an immunocompetent individual reflects, in part, a tumor escape from the immunosurveillance. The tumor escape is a complex, multifactorial, in which tumor cells will evade the defense mechanisms of the host by changing their microenvironment. Therefore, restoration or induction of these defense mechanisms is one of the therapeutic strategies against cancer. One of the principles of immunotherapy is based on the injection of antibodies that target tumor cells or effector cells of immunity. The anti-tumor efficacy of these antibodies has been greatly improved by a better understanding of modes of action and modulatory effects of these antibodies.Thus, to optimize the action of immune effectors to tumor cells, a bispecific antibody, trifunctional: catumaxomab, capable of binding to the adhesion molecule of the epithelial cells (EpCAM), expressed by tumor cells and the CD3 antigen of T cells, has been developed mainly in intraperitoneal treatment of refractory malignant ascites.The objective of this study was to determine the immunomodulatory effects of catumaxomab on tumoral cells expressing EpCAM, from two experimental models (allogeneic and autologous), evaluate and characterize cytotoxicity induced by catumaxomab, and analyze the presence of stress signals inducing immunogenic cell death such as membrane exposure of calreticulin by pre-apoptotic tumor cells, release of HMGB1 and of adenosine triphosphate (ATP) in the extracellular medium, inducing a T cell activation.In the presence of EpCAM + cells, catumaxomab induced a major the activation of T cells (expression of CD69, CD107a, HLA-DR and PD1), stimulated an inflammatory response Thelper type 1 (Th1) and the synthesis of interferon-gamma by CD8 T cells. Catumaxomab committed CD16 NK cells and monocytes. More, in models allogeneic catumaxomab, caused cell death associated with ATP release and induced an immunogenic cell death after pre-incubation of oxaliplatin.Therefore, catumaxomab modulates the immune environment in malignant ascites, and convert chronic and immunosuppressive inflammation (Th2) in acute and immunogenic inflammation (Th1). However, in these conditions, catumaxomab alone does not seem to trigger immunogenic cell death.the cytotoxicity of this bispecific antibody could be enhance by different techniques: (1) combining with chemotherapy such as oxaliplatin to promote immunogenic cell death, (2) refining its action on CD3 lymphocytes by changing its spatial configuration (BiTE antibody), (3) increasing its affinity for the FcR of accessory cells (Fc aglycosylated), (4) increasing its cytotoxicity by changing the target directed against the immune molecule (anti-PD-1 ...). Finally, the clinical use could be facilitated by this humanizing murine chimeric antibody to prevent the formation of anti-murine antibodies directed against catumaxomab.A phase II clinical trial aimed to evaluate the efficacy of intraperitoneal catumaxomab after complete cytoreductive surgery of gastric carcinomatosis in patients who received preoperative systemic chemotherapy with oxaliplatin have just started. In this study, we will validate the ability of catumaxomab 1) to induce immunogenic cell stress and death of cancer cells, 2) to change the polarization of effector cells to Th1 inflammatory disease, 3) to promote the expression of costimulatory molecules and TRAIL on NK cells and monocytes, and we will correlate these immune biomarkers to treatment efficacy.Le dĂ©veloppement d’un cancer chez un individu immunocompĂ©tent tĂ©moigne, en partie, d’un Ă©chappement tumoral au systĂšme d’immunosurveillance. Par consĂ©quent, la restauration ou l’induction de ces mĂ©canismes de dĂ©fense anti-tumorale est une des stratĂ©gies thĂ©rapeutiques actuelles. Un des principes de l’immunothĂ©rapie est basĂ© sur l’injection d’anticorps ayant pour cible la cellule tumorale ou les cellules effectrices de l’immunitĂ©. L’efficacitĂ© anti-tumorale de ces anticorps a Ă©tĂ© considĂ©rablement amĂ©liorĂ©e par une meilleure comprĂ©hension des modes d’action et des effets modulateurs de ces anticorps. Ainsi, afin d’optimiser l’action des effecteurs immunitaires sur les cellules tumorales, un anticorps bispĂ©cifique, trifonctionnel, le catumaxomab, capable de se lier Ă  la molĂ©cule d'adhĂ©rence des cellules Ă©pithĂ©liales (EpCAM) exprimĂ©e par les cellules tumorales et Ă  l'antigĂšne CD3 des lymphocytes T, a Ă©tĂ© dĂ©veloppĂ©, essentiellement en traitement intra-pĂ©ritonĂ©al des ascites nĂ©oplasiques rĂ©fractaires.L’objectif de cette Ă©tude Ă©tait de dĂ©terminer les effets immunomodulateurs du catumaxomab sur des cellules nĂ©oplasiques exprimant EpCAM, Ă  partir de deux modĂšles expĂ©rimentaux (allogĂ©nique et autologue), de rechercher une cytotoxicitĂ© induite par la catumaxomab, et de la caractĂ©riser, notamment en analysant la prĂ©sence ou non de signaux de stress inducteurs d’une mort immunogĂšne tels que l’exposition membranaire de la calrĂ©ticuline par les cellules tumorales prĂ©-apoptotiques, la libĂ©ration d’HMGB1 et d’adĂ©nosine triphosphate (ATP) dans le milieu extra-cellulaire, responsables d’une activation des lymphocytes T.En prĂ©sence de cellules EpCAM+, le catumaxomab entrainait une activation majeure des lymphocytes T (expression de CD69, CD107a, HLA-DR et PD1), stimulait une rĂ©ponse inflammatoire de type Thelper 1(Th1), et provoquait la synthĂšse d’interfĂ©ron-gamma par les lymphocytes T CD8. Le catumaxomab engageait le CD16 (FcR) des cellules monocytaires et NK. De plus, sur des modĂšles allogĂ©niques, le catumaxomab, provoquait une mort cellulaire associĂ©e Ă  la libĂ©ration d’ATP et induisait une mort immunogĂšne aprĂšs prĂ©-incubation dans de l’oxaliplatine.Par consĂ©quent, le catumaxomab permet de moduler l’environnement immunitaire dans les ascites nĂ©oplasiques, et de convertir une inflammation chronique et immunosuppressive (Th2) en une inflammation aigĂŒe et immunogĂšne (Th1). En revanche, dans ces conditions, l’administration seule de catumaxomab ne semble pas dĂ©clencher de mort immunogĂšne.DiffĂ©rents moyens pourraient permettre d’amĂ©liorer la cytotoxicitĂ© de cet anticorps bispĂ©cifique : (1) le combiner avec un agent anti-nĂ©oplasique tel que l’oxaliplatine afin de promouvoir une mort immunogĂšne, (2) affiner son action sur le CD3 des lymphocytes en modifiant sa configuration spatiale (anticorps BiTE), (3) amplifier son affinitĂ© pour le rĂ©cepteur Fcdes cellules accessoires (Fc dĂ©fucosylĂ©), (4) augmenter sa cytotoxicitĂ© en modifiant la cible dirigĂ©e contre la molĂ©cule du systĂšme immunitaire (anti-PD-1
). Enfin, l’utilisation clinique pourrait ĂȘtre facilitĂ©e en humanisant cet anticorps chimĂ©rique murin afin d’éviter la formation d’anticorps anti-murins, dirigĂ©s contre le catumaxomab.Un essai thĂ©rapeutique de phase II dont le but est d’évaluer l’efficacitĂ© du catumaxomab intrapĂ©ritonĂ©al aprĂšs chirurgie de cytorĂ©duction complĂšte d’une carcinose gastrique, chez des patients ayant reçu en prĂ©opĂ©ratoire une chimiothĂ©rapie systĂ©mique Ă  base d’oxaliplatine vient de dĂ©buter. Au cours de cette Ă©tude, nous allons valider la capacitĂ© du catumaxomab 1) Ă  induire un stress cellulaire immunogĂšne et la mort des cellules cancĂ©reuses, 2) Ă  modifier la polarisation des cellules effectrices vers une maladie inflammatoire Th1, 3) Ă  promouvoir l'expression des molĂ©cules de costimulation et TRAIL sur les cellules NK et monocytes, et corrĂ©ler ces biomarqueurs immunitaires Ă  l’efficacitĂ© du traitement
    • 

    corecore